Next Article in Journal
Vitamin D and Its Analogues Decrease Amyloid-β (Aβ) Formation and Increase Aβ-Degradation
Next Article in Special Issue
Using MOEA with Redistribution and Consensus Branches to Infer Phylogenies
Previous Article in Journal
Potent Inhibition of miR-34b on Migration and Invasion in Metastatic Prostate Cancer Cells by Regulating the TGF-β Pathway
Previous Article in Special Issue
Icariin Regulates Cellular Functions and Gene Expression of Osteoarthritis Patient-Derived Human Fibroblast-Like Synoviocytes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

A Genome-Wide Association Study and Complex Network Identify Four Core Hub Genes in Bipolar Disorder

Institute of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2017, 18(12), 2763; https://doi.org/10.3390/ijms18122763
Submission received: 23 October 2017 / Revised: 29 November 2017 / Accepted: 14 December 2017 / Published: 19 December 2017

Abstract

:
Bipolar disorder is a common and severe mental illness with unsolved pathophysiology. A genome-wide association study (GWAS) has been used to find a number of risk genes, but it is difficult for a GWAS to find genes indirectly associated with a disease. To find core hub genes, we introduce a network analysis after the GWAS was conducted. Six thousand four hundred fifty eight single nucleotide polymorphisms (SNPs) with p < 0.01 were sifted out from Wellcome Trust Case Control Consortium (WTCCC) dataset and mapped to 2045 genes, which are then compared with the protein–protein network. One hundred twelve genes with a degree >17 were chosen as hub genes from which five significant modules and four core hub genes (FBXL13, WDFY2, bFGF, and MTHFD1L) were found. These core hub genes have not been reported to be directly associated with BD but may function by interacting with genes directly related to BD. Our method engenders new thoughts on finding genes indirectly associated with, but important for, complex diseases.

Graphical Abstract

1. Introduction

Bipolar disorder (BD) is a common and severe mental disorder characterized by alternative episodes of mania/hypomania and depression [1]. It affects 1–5% of the world’s population [2,3,4]. Genetic studies have shown that bipolar disorder is a complex genetic disease that involves the interaction of multiple genes and the environment. Genetic factors can account for up to 60–85% of the risk [5,6,7,8]. The strong genetic basis of BD inspires plenty of research focused on finding candidate genes or single nucleotide polymorphisms (SNPs) associated with this disease.
Over the past few decades, traditional family-based linkage analysis and population-based case–control association analysis have been common means of identifying bipolar disorder susceptibility genes. With the advent of the third-generation polymorphism genetic marker SNPs, genome-wide association studies (GWASs) have also been applied to large-scale scanning of new BD susceptibility gene loci and a number of genes, such as CACAN1C [9,10], ANK3 [10,11], SYNE1 [12], CSMD1 [12], ITIH1 [11], KIT [11], and DGKH [13], have been found.
GWASs have proven to be useful in finding susceptibility genes of diseases. However, when used alone, it is difficult to determine genes that have relatively high GWAS p-values but may play a role through interaction with the genes directly associated with BD. The complexity of the disease makes it even more difficult to elucidate its molecular mechanism. Therefore, although the previous study has found a lot of genetic factors with significant effects on BD, its molecular mechanism remains unresolved. In this case, a comprehensive analysis focusing on gene interactions and biological functions will provide valuable information to explore the pathogenesis of BD. It has been found that the distribution of genetic marker loci on chromosomes and the interaction between SNPs are one of the major genetic basis for complex diseases [14]. The gene network is often used to reveal complex relationships among genes.
Considering that complex mental phenotypes may be affected by many genes with small or mild effects rather than one or two genes with a major impact [15], a comprehensive analysis of the underlying genes in the pathway or network framework may provide more insights into its molecular mechanism. It will be more efficient to understand the role of genes in complex diseases using network study. Some methods have been developed in this area, but the problem is far from being solved. There is scarce known molecular interaction mechanism and systematic gene network analysis for BD. Construction of a gene interaction network can be used to explore the synergistic effect of multiple genes on BD.
In this study, we performed a GWAS to obtain BD-related genes and confirmed their function by functional enrichment analysis. To further explore the association between these genes and BD, a network was constructed using a human protein–protein interaction database, and the BD-risk genes identified in the GWAS were mapped onto the network to find core hub genes. This will provide more insight into the molecular mechanisms of BD by determining the core hub genes of the network.

2. Results

2.1. GWAS Results

A total of 482,247 SNPs located on 22 chromosomes of 1868 BD cases and 2938 controls satisfies the quality control. The number of SNPs decreases to 354,282 after the Hardy–Weinberg equilibrium test. Finally, a total of 6458 SNPs is qualified in the GWAS where p < 0.01 and used for further analysis. The result is shown in Figure 1.

2.2. Gene Functional Analysis

A total of 2045 risk genes was obtained after mapping the 6458 SNPs onto human genes. These genes were then classified into three Gene Ontology (GO) sections: cellular components, molecular functions, and biological processes. The first 10 GO items (p < 0.01) are shown in Table 1, Table 2 and Table 3. Genes with transferase and kinase function dominate in molecular functions. In cellular components, most gene products are located in the nervous system. This coordinates with the biological process result in which most genes are involved in nervous system development.

2.3. Overlapped Genes in Different Mental Illnesses

We enriched these candidate genes in BD and other three mental illnesses: schizophrenia, intellectual disability, and autistic disorder. In the total 2045 risk genes, the numbers of genes associated with schizophrenia, intellectual disability, autism, and bipolar disorder are 151, 123, 84, and 84, respectively. A Venn map of the overlap genes of these four diseases shows that, out of the 84 genes associated with bipolar disorder, 55 genes are in common with schizophrenia, 17 with intellectual disorder, and 28 with autism (Figure 2).

2.4. Protein Interaction Network

The 2045 genes from the GWAS result are mapped onto the protein–protein interaction network constructed using data from the STRING database (Figure 3).
There are 1083 nodes in the network. The average node degree of the network is 7.555. The clustering coefficient is 0.232, and the characteristic path length is 3.393. The properties of the network are further analyzed and the results are shown in Figure 4. The connectivity of the network exhibits characteristic power distribution. Figure 4b shows that the shortest path with the highest frequency among the candidate genes of BD is between 3 and 4, indicating that the network is not a stochastic network but a complex network with characteristics of biological molecular network. The number of neighbors shared by the network nodes has a significant inverse relationship with its topological coefficients (Figure 4c), but shows a positive correlation with the node’s identity (Figure 4d).

2.5. Hub Genes of the Network

One hundred twelve gene nodes with a degree >17 is chosen as hub genes from the network for further analysis (Table 4 and Table A1).
Of these 112 hub genes, 45 were reported associated with BD in previous studies. Another 24 were reported associated with other mental illnesses. Gene nodes with higher degrees have a higher ratio of genes being reported associated with BD. Only five genes with a degree >23 (51 genes) are not reported associated with BD or other mental illnesses, while 24 with a degree ≤23 (61 genes) are not found reported directly associated to any mental illnesses. Obviously, risk genes with more degrees have a closer connection to BD than those with fewer degrees.

2.6. Significant Modules of the Network and Core Hub Genes

Five significant gene modules are found in the network containing 112 hub genes with Cytoscape. Four core hub genes are found in these modules: FBXL13, WDFY2, bFGF (FGF2), and MTHFD1L. No core hub gene is found for one module (Cluster 4) (Table 5, Figure 5).

3. Discussion

3.1. Most BD Risk Gene Products Are Located in the Nervous System

Our gene functional analysis of 2045 BD risk genes shows that most of their products are located in the nervous system, such as synapse and postsynapse. The result of GO biological process analysis shows most genes are involved in nervous system development. These two results verify each other and are consistent with previous studies [104]. BD risk genes may affect patients in two aspects: short-term and permanent. Environmental or internal factors may cause ectopic expression of some of the risk genes, which in turn cause episodes of BD. Some genes may work in the development of the nervous system and have a permanent effect on patients. This may explain why 60% patients will relapse into depression or mania within two years after treatment [105].

3.2. Intense Overlappings of Genes Associated with BD and Other Mental Disorders

Many symptoms and signs overlap between different mental disorders and patients often present with features of more than one disorder [106]. This may be caused by underlying genetic reasons. We compared BD risk genes with those of three other mental disorders and found intense overlaps. Similar results were also reported in other studies [28,52,86,89,106,107].
Schizophrenia and BD share the most associated genes. Previous work also found a significant correlation between a BP polygenic risk score and the clinical dimension of mania in schizophrenia patients [86]. PRKG1 was reported to be significantly associated with schizophrenia. In this study, we also find it is a hub gene in the network of BD risk genes. This gene encodes a cGMP-dependent protein kinase which acts as key mediator of the nitric oxide (NO)/cGMP signaling pathway. Another gene, SMARCA2, was also found to play a role in the pathophysiology of schizophrenia [27]. Its product is a transcription activator and involved in neuron differentiation. Many other risk genes are also found involved in signal transduction and nervous system development. This suggests that these two diseases may share some common underlying pathways.

3.3. Core Hub Genes Give New Insights of BD

We combined protein–protein network and genome wide association analysis in this study and found four core hub genes. Although genes with higher degrees are more frequently reported to be associated with BD, two core hub genes (WDFY2 and FBXL13) have relatively low degrees (20 and 19, respectively).
bFGF has not been reported to be associated with BD before, but is usually used for treatment of neurodegenerative diseases such as Alzheimer’s disease [42]. It plays an essential role in regulation of cell proliferation, differentiation, and migration. bFGF is found as a core hub gene implies the abnormal nervous development of BD patients.
There is no obvious evidence for another core hub gene, MTHFD1L, to be associated with BD, but it is thought to have an important effect on the pathophysiology of depression through rumination, and maybe via this cognitive intermediate phenotype on other mental and physical disorders [38].
WDFY2 is not directly associated with BD, but its product interacts with AKT1 [108], which has been found involved in BD and schizophrenia [109]. This result suggests that the pathophysiology of BD is even more complicated than we thought. Some genes may play a role through its interaction with genes directly associated with BD.
FBXL13 functions in the maturation of human dendritic cells [68] which are key regulators in the immune system and show mild aberrancies in bipolar disorder that can be fully restored to even activation after in vivo lithium treatment [67].
Interestingly, all the four core hub genes are not directly associated with BD. Although the role of these genes in the pathophysiology of BD requires further investigation, our method inspires new initiatives to find those genes that are important for BD but overlooked by studies using GWAS alone.

3.4. Effectiveness of GWAS Followed by Gene Network Analysis

GWAS is a successful tool for identifying human disease-associated genes. However, results of different studies often vary due to sampling even when a strict significant p-value of 5 × 10−8 is used [110]. In this study, a loose p-value threshold of 0.01 was used for the GWAS, and a gene network analysis was then used to find BD-associated genes in the GWAS result. Many resulted genes with high network degrees but relatively high GWAS p-values are reported to be associated with BD and/or other mental illnesses (Table 4), which suggests that the combination of the two methods is efficient in finding disease-related genes. It is necessary to use a loose p-value threshold in the first step to provide enough input genes for the following network analysis. A second screening using network degrees can help to make the final result more reliable.
Sklar et al. conducted a combined GWAS with 7481 BD cases and 9250 controls and identified CACNA1C and a miRNA located in the first intron of ODZ4 as BD-associated genes [87]. The calcium channel subunit coding gene CACNA1C has also been found to be associated with BD in previous studies [9,52,86] and is confirmed with a relatively high degree (30) in our results. However, the miRNA is not detected in this study, probably due to our relatively smaller sample size.

4. Materials and Methods

4.1. Bipolar Disorder Datasets

The dataset is from a study published by Wellcome Trust Case Control Consortium (WTCCC), which conducted a genome-wide scan of all SNPs of 17,000 British Caucasian loci by human SNPs genotyping chips. This dataset includes 14,000 disease samples from seven common complex diseases: bipolar disorder, bipolar depression, Crohn’s disease, hypertension, rheumatoid arthritis, type 1 diabetes, type 2 diabetes, and 3000 healthy control samples, which has been completed by more than 50 research teams [111]. The dataset is downloaded from WTCCC website [112]. This study uses the BD part of the dataset. Human SNP annotation data and human reference sequence data are downloaded from NCBI (https://www.ncbi.nlm.nih.gov/), which contain 336,843,011 SNPs on 24 human chromosomes and the start and end of genes in which they are located [113].

4.2. Screening of Risk SNPs

SNP sites that do not meet one of the following criteria are excluded for quality control: Hardy–Weinberg equilibrium test (Bonferroni corrected p < 5 × 10−7), missingness >5%, minor allele frequency <5%, and odds ratio R2 > 0.8. Risk SNPs are screened under p < 0.01. Quality control and risk gene screening are finished with Plink software [114].

4.3. Mapping Significant Risk SNPs to Genes

Risk SNPs are mapped onto human genes by comparing them with transcription start sites and stop sites. An SNP will be mapped onto its nearest gene within 5 kb if it is not located within any gene. SNPs located outside of the 5 kb of genes are removed.

4.4. Gene Function and Disease Enrichment Analysis

FunRich [115] software is used to carry out gene enrichment analysis with p < 0.01. Results are reversely ordered by FDR-values, and only the first 10 results are listed in each GO section. ToppGene [116] is used to enrich genes in four different mental illnesses.

4.5. Protein Network Analysis

STRING database [117] is used to find a protein–protein relationship and FunRich is then used to map BD risk genes to the protein–protein network. Those protein (gene) nodes with degree >17 are sifted out as hub genes, which are further analyzed with the MCODE plugin [118] of Cytoscape [119] to find out network clusters (modules) and core hub genes. The node gene with the highest MCODE node score in a cluster is designated as its core hub gene, which is crucial for the cluster.
The topological properties of a gene cluster include [120,121] the following: (1) degree, the number of genes directly connected to a gene, (2) the cluster coefficient (CC), the coincidence of the common regulatory genes between two adjacent genes, defined as
CC = 2 n i k i ( k i 1 )
where n i represents the number of edges of the k i neighbors that connect to node i —the mean of the clustering coefficients of all nodes is designated as the clustering coefficient of the network—(3) the shortest path, the path with the least edges between two nodes, and (4) betweenness (B(v)), the sum of the ratios of number of shortest paths connecting to a node to that of all shortest paths in a network
B ( ν ) = s ν , s t , ν t ν δ s t ( ν ) δ s t
where δ s t is total number of shortest paths from node s to t, and δ s t ( ν ) is the number of those paths that pass through v.

Acknowledgments

This study was financially supported by the Special Project of National Science and Technology Cooperation (2014DFB30010), National Natural Science Foundation of China (61501071) and the Science and Technology Research Program of Chongqing Municipal Education Commission (KJ1704094). We thank the three anonymous reviewers for their constructive comments.

Author Contributions

Zengyan Xie contributed literature search, study design, data interpretation, and wrote the paper and provided study supervision. Xianyan Yang contributed literature search, figures, study design, data collection, data analysis, data interpretation and wrote the paper. Xiaoya Deng contributed literature search and data checking. Mingyue Ma contributed paper revising. Kunxian Shu contributed study design and provided study supervision.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

BDbipolar disorder
GWASgenome-wide association study
SNPsingle nucleotide polymorphism
WTCCCthe World Healthcare Case Control Association

Appendix A

Table A1. Gene information of the nodes with a network degree greater than 17.
Table A1. Gene information of the nodes with a network degree greater than 17.
GENEDegreeEnsemblUniProtKB
CDK162ENSG00000170312P06493
PTEN61ENSG00000171862P60484
BCL260ENSG00000171791P10415
POLR2A55ENSG00000181222P24928
SMARCA255ENSG00000080503P5153
GSK3B54ENSG00000082701P49841
ABL153ENSG00000097007P00519
PRKCA50ENSG00000154229P17252
FGF248ENSG00000138685P09038
RB145ENSG00000139687P06400
KIT40ENSG00000157404P10721
RAD5138ENSG00000051180Q06609
SIRT138ENSG00000096717Q96EB6
UBE2D137ENSG00000072401P51668
DLG136ENSG00000075711Q12959
CDC2735ENSG00000004897P30260
NEDD4L35ENSG00000049759Q96PU5
PRKG135ENSG00000185532Q13976
RAP1A34ENSG00000116473P62834
CDH233ENSG00000170558P19022
GNB533ENSG00000069966O14775
MAPK633ENSG00000069956Q16659
GNG732ENSG00000176533O60262
PTPN1132ENSG00000179295Q06124
ZBTB1632ENSG00000109906Q05516
ADCY831ENSG00000155897P40145
DICER131ENSG00000100697Q9UPY3
SYNJ131ENSG00000159082O43426
CACNA1C30ENSG00000151067Q13936
CTTN30ENSG00000085733Q14247
DLG230ENSG00000150672Q15700
MAP3K130ENSG00000095015Q13233
RIT230ENSG00000152214Q99578
ANAPC528ENSG00000089053Q9UJX4
PLCB128ENSG00000182621Q9NQ66
RAF128ENSG00000132155P04049
PARK227ENSG00000185345O60260
PLCG227ENSG00000197943P16885
PNPLA627ENSG00000032444Q8IY17
SYNJ227ENSG00000078269O15056
UBE2R227ENSG00000107341Q712K3
CACNA1D26ENSG00000157388Q01668
CDK626ENSG00000105810Q00534
CHRM226ENSG00000181072P08172
MTHFD1L26ENSG00000120254Q6UB35
GRIA125ENSG00000155511P42261
POLR2H25ENSG00000163882P52434
TJP125ENSG00000104067Q07157
MAPRE124ENSG00000101367Q15691
RUNX124ENSG00000159216Q01196
UBE2D424ENSG00000078967Q9Y2X8
EHHADH23ENSG00000113790Q08426
IQCB123ENSG00000173226Q15051
PPM1B23ENSG00000138032O75688
PPP4C23ENSG00000149923P60510
RAD5023ENSG00000113522Q92878
SH3GL223ENSG00000107295Q99962
DCTN122ENSG00000204843Q14203
ERBB422ENSG00000178568Q15303
FBXO3222ENSG00000156804Q969P5
ITPR122ENSG00000150995Q14643
MLL22ENSG00000118058Q03164
NCOR222ENSG00000196498Q9Y618
PRKCE22ENSG00000171132Q02156
RAD51B22ENSG00000182185O15315
ACTN421ENSG00000130402O43707
CCND221ENSG00000118971P30279
CDH521ENSG00000179776P33151
CUL4A21ENSG00000139842Q13619
EFCAB1321ENSG00000178852Q8IY85
LMO721ENSG00000136153Q8WWI1
MITF21ENSG00000187098O75030
TRIM921ENSG00000100505Q9C026
CCND320ENSG00000112576P30281
EPHB120ENSG00000154928P54762
FARS220ENSG00000145982O95363
FBXO2220ENSG00000167196Q8NEZ5
FLT320ENSG00000122025P36888
GATA420ENSG00000136574P43694
ITSN220ENSG00000198399Q9NZM3
KIF18A20ENSG00000121621Q8NI77
LONRF120ENSG00000154359Q17RB8
NCOA320ENSG00000124151Q9Y6Q9
PCNT20ENSG00000160299O95613
PJA220ENSG00000198961O43164
SYT120ENSG00000067715P21579
TRIM3920ENSG00000204599Q9HCM9
WDFY220ENSG00000139668Q96P53
AK419ENSG00000162433P27144
ASB1519ENSG00000146809Q8WXK1
ATF219ENSG00000115966P15336
BUB1B19ENSG00000156970O60566
DHX1519ENSG00000109606O43143
DNM319ENSG00000197959Q9UQ16
ETV619ENSG00000139083P41212
FBXL1319ENSG00000161040Q8NEE6
HECW219ENSG00000138411Q9P2P5
MEF2C19ENSG00000081189Q06413
NR3C119ENSG00000113580P04150
PDE4D19ENSG00000113448Q08499
RNF19B19ENSG00000116514Q6ZMZ0
RNF21719ENSG00000146373Q8TC41
RXFP219ENSG00000133105Q8WXD0
RYR119ENSG00000196218P21817
THBS219ENSG00000186340P35442
AKT318ENSG00000117020Q9Y243
BARD118ENSG00000138376Q99728
CTNNA218ENSG00000066032P26232
HDAC718ENSG00000061273Q8WUI4
ITGAV18ENSG00000138448P06756
PARD318ENSG00000148498Q8TEW0
PCSK218ENSG00000125851P16519
PIK3C2B18ENSG00000133056O00750
PIK3C2G18ENSG00000139144O75747
UBQLN118ENSG00000135018Q9UMX0

References

  1. Craddock, N.; Sklar, P. Genetics of bipolar disorder. Lancet 2013, 381, 1654–1662. [Google Scholar] [CrossRef]
  2. Akiskal, H.S.; Bourgeois, M.L.; Angst, J.; Post, R.; Moller, H.; Hirschfeld, R. Re-evaluating the prevalence of and diagnostic composition within the broad clinical spectrum of bipolar disorders. J. Affect. Disord. 2000, 59, S5–S30. [Google Scholar] [CrossRef]
  3. Kessler, R.C.; Akiskal, H.S.; Ames, M.; Birnbaum, H.; Greenberg, P.; Hirschfeld, R.M.; Jin, R.; Merikangas, K.R.; Simon, G.E.; Wang, P.S. Prevalence and effects of mood disorders on work performance in a nationally representative sample of U.S. workers. Am. J. Psychiatry 2006, 163, 1561–1568. [Google Scholar] [CrossRef] [PubMed]
  4. Grande, I.; Berk, M.; Birmaher, B.; Vieta, E. Bipolar disorder. Lancet 2016, 387, 1561–1572. [Google Scholar] [CrossRef]
  5. Smoller, J.W.; Finn, C.T. Family, twin, and adoption studies of bipolar disorder. Am. J. Med. Genet. Part C Semin. Med. Genet. 2003, 123C, 48–58. [Google Scholar] [CrossRef] [PubMed]
  6. Barnett, J.H.; Smoller, J.W. The genetics of bipolar disorder. Neuroscience 2009, 164, 331–343. [Google Scholar] [CrossRef] [PubMed]
  7. Sullivan, P.F.; Daly, M.J.; O’Donovan, M. Genetic architectures of psychiatric disorders: The emerging picture and its implications. Nat. Rev. Genet. 2012, 13, 537–551. [Google Scholar] [CrossRef] [PubMed]
  8. Lescai, F.; Als, T.D.; Li, Q.; Nyegaard, M.; Andorsdottir, G.; Biskopsto, M.; Hedemand, A.; Fiorentino, A.; O’Brien, N.; Jarram, A.; et al. Whole-exome sequencing of individuals from an isolated population implicates rare risk variants in bipolar disorder. Transl. Psychiatry 2017, 7, e1034. [Google Scholar] [CrossRef] [PubMed]
  9. Sklar, P.; Smoller, J.W.; Fan, J.; Ferreira, M.A.; Perlis, R.H.; Chambert, K.; Nimgaonkar, V.L.; McQueen, M.B.; Faraone, S.V.; Kirby, A.; et al. Whole-genome association study of bipolar disorder. Mol. Psychiatry 2008, 13, 558–569. [Google Scholar] [CrossRef] [PubMed]
  10. Ferreira, M.A.; O’Donovan, M.C.; Meng, Y.A.; Jones, I.R.; Ruderfer, D.M.; Jones, L.; Fan, J.; Kirov, G.; Perlis, R.H.; Green, E.K.; et al. Collaborative genome-wide association analysis supports a role for ANK3 and CACNA1C in bipolar disorder. Nat. Genet. 2008, 40, 1056–1058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Scott, L.J.; Muglia, P.; Kong, X.Q.; Guan, W.; Flickinger, M.; Upmanyu, R.; Tozzi, F.; Li, J.Z.; Burmeister, M.; Absher, D.; et al. Genome-wide association and meta-analysis of bipolar disorder in individuals of European ancestry. Proc. Natl. Acad. Sci. USA 2009, 106, 7501–7506. [Google Scholar] [CrossRef] [PubMed]
  12. Xu, W.; Cohen-Woods, S.; Chen, Q.; Noor, A.; Knight, J.; Hosang, G.; Parikh, S.V.; De Luca, V.; Tozzi, F.; Muglia, P.; et al. Genome-wide association study of bipolar disorder in Canadian and UK populations corroborates disease loci including SYNE1 and CSMD1. BMC Med. Genet. 2014, 15, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Baum, A.E.; Akula, N.; Cabanero, M.; Cardona, I.; Corona, W.; Klemens, B.; Schulze, T.G.; Cichon, S.; Rietschel, M.; Nothen, M.M.; et al. A genome-wide association study implicates diacylglycerol kinase eta (DGKH) and several other genes in the etiology of bipolar disorder. Mol. Psychiatry 2008, 13, 197–207. [Google Scholar] [CrossRef] [PubMed]
  14. Martin, N.W.; Medland, S.E.; Verweij, K.J.; Lee, S.H.; Nyholt, D.R.; Madden, P.A.; Heath, A.C.; Montgomery, G.W.; Wright, M.J.; Martin, N.G. Educational attainment: A genome wide association study in 9538 Australians. PLoS ONE 2011, 6, e20128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. McGuffin, P.; Rijsdijk, F.; Andrew, M.; Sham, P.; Katz, R.; Cardno, A. The heritability of bipolar affective disorder and the genetic relationship to unipolar depression. Arch. Gen. Psychiatry 2003, 60, 497–502. [Google Scholar] [CrossRef] [PubMed]
  16. Song, Y.; Wang, M.; Mao, F.; Shao, M.; Zhao, B.; Song, Z.; Shao, C.; Gong, Y. Knockdown of Pnpla6 protein results in motor neuron defects in zebrafish. Dis. Models Mech. 2013, 6, 404–413. [Google Scholar] [CrossRef] [PubMed]
  17. Le-Niculescu, H.; Levey, D.F.; Ayalew, M.; Palmer, L.; Gavrin, L.M.; Jain, N.; Winiger, E.; Bhosrekar, S.; Shankar, G.; Radel, M.; et al. Discovery and validation of blood biomarkers for suicidality. Mol. Psychiatry 2013, 18, 1249–1264. [Google Scholar] [CrossRef] [PubMed]
  18. Quintero-Rivera, F.; Sharifi-Hannauer, P.; Martinez-Agosto, J.A. Autistic and psychiatric findings associated with the 3q29 microdeletion syndrome: Case report and review. Am. J. Med. Genet. Part A 2010, 152A, 2459–2467. [Google Scholar] [CrossRef] [PubMed]
  19. Uemura, T.; Green, M.; Corson, T.W.; Perova, T.; Li, P.P.; Warsh, J.J. Bcl-2 SNP rs956572 associates with disrupted intracellular calcium homeostasis in bipolar I disorder. Bipolar Disord. 2011, 13, 41–51. [Google Scholar] [CrossRef] [PubMed]
  20. Zhernakova, D.V.; de Klerk, E.; Westra, H.J.; Mastrokolias, A.; Amini, S.; Ariyurek, Y.; Jansen, R.; Penninx, B.W.; Hottenga, J.J.; Willemsen, G.; et al. DeepSAGE reveals genetic variants associated with alternative polyadenylation and expression of coding and non-coding transcripts. PLoS Genet. 2013, 9, e1003594. [Google Scholar] [CrossRef]
  21. Silberberg, G.; Baruch, K.; Navon, R. Detection of stable reference genes for real-time PCR analysis in schizophrenia and bipolar disorder. Anal. Biochem. 2009, 391, 91–97. [Google Scholar] [CrossRef] [PubMed]
  22. Ross, J.; Gedvilaite, E.; Badner, J.A.; Erdman, C.; Baird, L.; Matsunami, N.; Leppert, M.; Xing, J.; Byerley, W. A Rare Variant in CACNA1D Segregates with 7 Bipolar I Disorder Cases in a Large Pedigree. Mol. Neuropsychiatry 2016, 2, 145–150. [Google Scholar] [CrossRef] [PubMed]
  23. Kabir, Z.D.; Martinez-Rivera, A.; Rajadhyaksha, A.M. From Gene to Behavior: L-Type Calcium Channel Mechanisms Underlying Neuropsychiatric Symptoms. Neurotherapeutics 2017, 14, 588–613. [Google Scholar] [CrossRef] [PubMed]
  24. Martinez-Rivera, A.; Hao, J.; Tropea, T.F.; Giordano, T.P.; Kosovsky, M.; Rice, R.C.; Lee, A.; Huganir, R.L.; Striessnig, J.; Addy, N.A.; et al. Enhancing VTA Cav1.3 L-type Ca2+ channel activity promotes cocaine and mood-related behaviors via overlapping AMPA receptor mechanisms in the nucleus accumbens. Mol. Psychiatry 2017, 22, 1735–1745. [Google Scholar] [CrossRef] [PubMed]
  25. Teschler, S.; Bartkuhn, M.; Kunzel, N.; Schmidt, C.; Kiehl, S.; Dammann, G.; Dammann, R. Aberrant methylation of gene associated CpG sites occurs in borderline personality disorder. PLoS ONE 2013, 8, e84180. [Google Scholar] [CrossRef] [PubMed]
  26. Sengupta, S.; Xiong, L.; Fathalli, F.; Benkelfat, C.; Tabbane, K.; Danics, Z.; Labelle, A.; Lal, S.; Krebs, M.O.; Rouleau, G.; et al. Association study of the trinucleotide repeat polymorphism within SMARCA2 and schizophrenia. BMC Genet. 2006, 7, 34. [Google Scholar] [CrossRef] [PubMed]
  27. Koga, M.; Ishiguro, H.; Yazaki, S.; Horiuchi, Y.; Arai, M.; Niizato, K.; Iritani, S.; Itokawa, M.; Inada, T.; Iwata, N.; et al. Involvement of SMARCA2/BRM in the SWI/SNF chromatin-remodeling complex in schizophrenia. Hum. Mol. Genet. 2009, 18, 2483–2494. [Google Scholar] [CrossRef] [PubMed]
  28. Benes, F.M.; Lim, B.; Subburaju, S. Site-specific regulation of cell cycle and DNA repair in post-mitotic GABA cells in schizophrenic versus bipolars. Proc. Natl. Acad. Sci. USA 2009, 106, 11731–11736. [Google Scholar] [CrossRef] [PubMed]
  29. Vine, A.E.; McQuillin, A.; Bass, N.J.; Pereira, A.; Kandaswamy, R.; Robinson, M.; Lawrence, J.; Anjorin, A.; Sklar, P.; Gurling, H.M.; et al. No evidence for excess runs of homozygosity in bipolar disorder. Psychiatr. Genet. 2009, 19, 165–170. [Google Scholar] [CrossRef] [PubMed]
  30. Benedetti, F.; Serretti, A.; Colombo, C.; Lorenzi, C.; Tubazio, V.; Smeraldi, E. A glycogen synthase kinase 3-beta promoter gene single nucleotide polymorphism is associated with age at onset and response to total sleep deprivation in bipolar depression. Neurosci. Lett. 2004, 368, 123–126. [Google Scholar] [CrossRef] [PubMed]
  31. Hu, S.; Begum, A.N.; Jones, M.R.; Oh, M.S.; Beech, W.K.; Beech, B.H.; Yang, F.; Chen, P.; Ubeda, O.J.; Kim, P.C.; et al. GSK3 inhibitors show benefits in an Alzheimer’s disease (AD) model of neurodegeneration but adverse effects in control animals. Neurobiol. Dis. 2009, 33, 193–206. [Google Scholar] [CrossRef] [PubMed]
  32. Li, X.; Jope, R.S. Is glycogen synthase kinase-3 a central modulator in mood regulation? Neuropsychopharmacology 2010, 35, 2143–2154. [Google Scholar] [CrossRef] [PubMed]
  33. Benedetti, F.; Bollettini, I.; Barberi, I.; Radaelli, D.; Poletti, S.; Locatelli, C.; Pirovano, A.; Lorenzi, C.; Falini, A.; Colombo, C.; et al. Lithium and GSK3-beta promoter gene variants influence white matter microstructure in bipolar disorder. Neuropsychopharmacology 2013, 38, 313–327. [Google Scholar] [CrossRef] [PubMed]
  34. Cannon, D.M.; Klaver, J.K.; Gandhi, S.K.; Solorio, G.; Peck, S.A.; Erickson, K.; Akula, N.; Savitz, J.; Eckelman, W.C.; Furey, M.L.; et al. Genetic variation in cholinergic muscarinic-2 receptor gene modulates M2 receptor binding in vivo and accounts for reduced binding in bipolar disorder. Mol. Psychiatry 2011, 16, 407–418. [Google Scholar] [CrossRef] [PubMed]
  35. Munkholm, K.; Peijs, L.; Vinberg, M.; Kessing, L.V. A composite peripheral blood gene expression measure as a potential diagnostic biomarker in bipolar disorder. Transl. Psychiatry 2015, 5, e614. [Google Scholar] [CrossRef] [PubMed]
  36. Kato, T.; Hayashi-Takagi, A.; Toyota, T.; Yoshikawa, T.; Iwamoto, K. Gene expression analysis in lymphoblastoid cells as a potential biomarker of bipolar disorder. J. Hum. Genet. 2011, 56, 779–783. [Google Scholar] [CrossRef] [PubMed]
  37. Munkholm, K.; Peijs, L.; Kessing, L.V.; Vinberg, M. Reduced mRNA expression of PTGDS in peripheral blood mononuclear cells of rapid-cycling bipolar disorder patients compared with healthy control subjects. Int. J. Neuropsychopharmacol. 2014, 18. [Google Scholar] [CrossRef] [PubMed]
  38. Eszlari, N.; Kovacs, D.; Petschner, P.; Pap, D.; Gonda, X.; Elliott, R.; Anderson, I.M.; Deakin, J.F.; Bagdy, G.; Juhasz, G. Distinct effects of folate pathway genes MTHFR and MTHFD1L on ruminative response style: A potential risk mechanism for depression. Transl. Psychiatry 2016, 6, e745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Kittel-Schneider, S.; Lorenz, C.; Auer, J.; Weissflog, L.; Reif, A. DGKH genetic risk variant influences gene expression in bipolar affective disorder. J. Affect. Disord. 2016, 198, 148–157. [Google Scholar] [CrossRef] [PubMed]
  40. Carroll, L.S.; Williams, N.M.; Moskvina, V.; Russell, E.; Norton, N.; Williams, H.J.; Peirce, T.; Georgieva, L.; Dwyer, S.; Grozeva, D.; et al. Evidence for rare and common genetic risk variants for schizophrenia at protein kinase C, alpha. Mol. Psychiatry 2010, 15, 1101–1111. [Google Scholar] [CrossRef] [PubMed]
  41. Kerner, B.; Jasinska, A.J.; DeYoung, J.; Almonte, M.; Choi, O.W.; Freimer, N.B. Polymorphisms in the GRIA1 gene region in psychotic bipolar disorder. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 2009, 150B, 24–32. [Google Scholar] [CrossRef] [PubMed]
  42. Feng, C.; Zhang, C.; Shao, X.; Liu, Q.; Qian, Y.; Feng, L.; Chen, J.; Zha, Y.; Zhang, Q.; Jiang, X. Enhancement of nose-to-brain delivery of basic fibroblast growth factor for improving rat memory impairments induced by co-injection of beta-amyloid and ibotenic acid into the bilateral hippocampus. Int. J. Pharm. 2012, 423, 226–234. [Google Scholar] [CrossRef] [PubMed]
  43. Numata, S.; Iga, J.; Nakataki, M.; Tayoshi, S.; Tanahashi, T.; Itakura, M.; Ueno, S.; Ohmori, T. Positive association of the pericentrin (PCNT) gene with major depressive disorder in the Japanese population. J. Psychiatry Neurosci. 2009, 34, 195–198. [Google Scholar] [PubMed]
  44. De Ligt, J.; Willemsen, M.H.; van Bon, B.W.; Kleefstra, T.; Yntema, H.G.; Kroes, T.; Vulto-van Silfhout, A.T.; Koolen, D.A.; de Vries, P.; Gilissen, C.; et al. Diagnostic exome sequencing in persons with severe intellectual disability. N. Engl. J. Med. 2012, 367, 1921–1929. [Google Scholar] [CrossRef] [PubMed]
  45. Iossifov, I.; Zheng, T.; Baron, M.; Gilliam, T.C.; Rzhetsky, A. Genetic-linkage mapping of complex hereditary disorders to a whole-genome molecular-interaction network. Genome Res. 2008, 18, 1150–1162. [Google Scholar] [CrossRef] [PubMed]
  46. Drago, A.; Crisafulli, C.; Sidoti, A.; Calabro, M.; Serretti, A. The microtubule-associated molecular pathways may be genetically disrupted in patients with Bipolar Disorder. Insights from the molecular cascades. J. Affect. Disord. 2016, 190, 429–438. [Google Scholar] [CrossRef] [PubMed]
  47. Cupertino, R.B.; Kappel, D.B.; Bandeira, C.E.; Schuch, J.B.; da Silva, B.S.; Muller, D.; Bau, C.H.; Mota, N.R. SNARE complex in developmental psychiatry: Neurotransmitter exocytosis and beyond. J. Neural Transm. 2016, 123, 867–883. [Google Scholar] [CrossRef] [PubMed]
  48. Serretti, A.; Mandelli, L. The genetics of bipolar disorder: Genome ‘hot regions’, genes, new potential candidates and future directions. Mol. Psychiatry 2008, 13, 742–771. [Google Scholar] [CrossRef] [PubMed]
  49. Kishi, T.; Fukuo, Y.; Kitajima, T.; Okochi, T.; Yamanouchi, Y.; Kinoshita, Y.; Kawashima, K.; Inada, T.; Kunugi, H.; Kato, T.; et al. SIRT1 gene, schizophrenia and bipolar disorder in the Japanese population: An association study. Genes Brain Behav. 2011, 10, 257–263. [Google Scholar] [CrossRef] [PubMed]
  50. Herskovits, A.Z.; Guarente, L. SIRT1 in neurodevelopment and brain senescence. Neuron 2014, 81, 471–483. [Google Scholar] [CrossRef] [PubMed]
  51. Nivoli, A.; Porcelli, S.; Albani, D.; Forloni, G.; Fusco, F.; Colom, F.; Vieta, E.; Serretti, A. Association between Sirtuin 1 Gene rs10997870 Polymorphism and Suicide Behaviors in Bipolar Disorder. Neuropsychobiology 2016, 74, 1–7. [Google Scholar] [CrossRef] [PubMed]
  52. Moskvina, V.; Craddock, N.; Holmans, P.; Nikolov, I.; Pahwa, J.S.; Green, E.; Wellcome Trust Case Control Consortium; Owen, M.J.; O’Donovan, M.C. Gene-wide analyses of genome-wide association data sets: Evidence for multiple common risk alleles for schizophrenia and bipolar disorder and for overlap in genetic risk. Mol. Psychiatry 2009, 14, 252–260. [Google Scholar] [CrossRef] [PubMed]
  53. Smalheiser, N.R.; Lugli, G.; Rizavi, H.S.; Torvik, V.I.; Turecki, G.; Dwivedi, Y. MicroRNA expression is down-regulated and reorganized in prefrontal cortex of depressed suicide subjects. PLoS ONE 2012, 7, e33201. [Google Scholar] [CrossRef] [PubMed]
  54. Dwivedi, Y. Emerging role of microRNAs in major depressive disorder: Diagnosis and therapeutic implications. Dialogues Clin. Neurosci. 2014, 16, 43–61. [Google Scholar] [PubMed]
  55. Uezato, A.; Yamamoto, N.; Iwayama, Y.; Hiraoka, S.; Hiraaki, E.; Umino, A.; Haramo, E.; Umino, M.; Yoshikawa, T.; Nishikawa, T. Reduced cortical expression of a newly identified splicing variant of the DLG1 gene in patients with early-onset schizophrenia. Transl. Psychiatry 2015, 5, e654. [Google Scholar] [CrossRef] [PubMed]
  56. Xing, J.; Kimura, H.; Wang, C.; Ishizuka, K.; Kushima, I.; Arioka, Y.; Yoshimi, A.; Nakamura, Y.; Shiino, T.; Oya-Ito, T.; et al. Resequencing and Association Analysis of Six PSD-95-Related Genes as Possible Susceptibility Genes for Schizophrenia and Autism Spectrum Disorders. Sci. Rep. 2016, 6, 27491. [Google Scholar] [CrossRef] [PubMed]
  57. Hunsberger, J.G.; Chibane, F.L.; Elkahloun, A.G.; Henderson, R.; Singh, R.; Lawson, J.; Cruceanu, C.; Nagarajan, V.; Turecki, G.; Squassina, A.; et al. Novel integrative genomic tool for interrogating lithium response in bipolar disorder. Transl. Psychiatry 2015, 5, e504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Le-Niculescu, H.; Patel, S.D.; Bhat, M.; Kuczenski, R.; Faraone, S.V.; Tsuang, M.T.; McMahon, F.J.; Schork, N.J.; Nurnberger, J.I., Jr.; Niculescu, A.B., 3rd. Convergent functional genomics of genome-wide association data for bipolar disorder: Comprehensive identification of candidate genes, pathways and mechanisms. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 2009, 150B, 155–181. [Google Scholar] [CrossRef] [PubMed]
  59. Chen, H.; Ross, C.A.; Wang, N.; Huo, Y.; MacKinnon, D.F.; Potash, J.B.; Simpson, S.G.; McMahon, F.J.; DePaulo, J.R., Jr.; McInnis, M.G. NEDD4L on human chromosome 18q21 has multiple forms of transcripts and is a homologue of the mouse Nedd4-2 gene. Eur. J. Hum. Genet. 2001, 9, 922–930. [Google Scholar] [CrossRef] [PubMed]
  60. Birnbaum, R.; Jaffe, A.E.; Hyde, T.M.; Kleinman, J.E.; Weinberger, D.R. Prenatal expression patterns of genes associated with neuropsychiatric disorders. Am. J. Psychiatry 2014, 171, 758–767. [Google Scholar] [CrossRef] [PubMed]
  61. Zhao, Z.; Webb, B.T.; Jia, P.; Bigdeli, T.B.; Maher, B.S.; van den Oord, E.; Bergen, S.E.; Amdur, R.L.; O’Neill, F.A.; Walsh, D.; et al. Association study of 167 candidate genes for schizophrenia selected by a multi-domain evidence-based prioritization algorithm and neurodevelopmental hypothesis. PLoS ONE 2013, 8, e67776. [Google Scholar] [CrossRef] [PubMed]
  62. Sakai, M.; Watanabe, Y.; Someya, T.; Araki, K.; Shibuya, M.; Niizato, K.; Oshima, K.; Kunii, Y.; Yabe, H.; Matsumoto, J.; et al. Assessment of copy number variations in the brain genome of schizophrenia patients. Mol. Cytogenet. 2015, 8, 46. [Google Scholar] [CrossRef] [PubMed]
  63. Moya, P.R.; Dodman, N.H.; Timpano, K.R.; Rubenstein, L.M.; Rana, Z.; Fried, R.L.; Reichardt, L.F.; Heiman, G.A.; Tischfield, J.A.; King, R.A.; et al. Rare missense neuronal cadherin gene (CDH2) variants in specific obsessive-compulsive disorder and Tourette disorder phenotypes. Eur. J. Hum. Genet. (EJHG) 2013, 21, 850–854. [Google Scholar] [CrossRef] [PubMed]
  64. Lodder, E.M.; De Nittis, P.; Koopman, C.D.; Wiszniewski, W.; Moura de Souza, C.F.; Lahrouchi, N.; Guex, N.; Napolioni, V.; Tessadori, F.; Beekman, L.; et al. GNB5 Mutations Cause an Autosomal-Recessive Multisystem Syndrome with Sinus Bradycardia and Cognitive Disability. Am. J. Hum. Genet. 2016, 99, 704–710. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, P.; Chen, J.; Huang, K.; Ji, W.; Wang, T.; Li, T.; Wang, Y.; Wang, H.; He, L.; Feng, G.; et al. Analysis of association between common SNPs in ErbB4 and bipolar affective disorder, major depressive disorder and schizophrenia in the Han Chinese population. Prog. Neuro-psychopharmacol. Biol. Psychiatry 2012, 36, 17–21. [Google Scholar] [CrossRef] [PubMed]
  66. Goes, F.S.; Rongione, M.; Chen, Y.C.; Karchin, R.; Elhaik, E.; Bipolar Genome, S.; Potash, J.B. Exonic DNA sequencing of ERBB4 in bipolar disorder. PLoS ONE 2011, 6, e20242. [Google Scholar] [CrossRef] [PubMed]
  67. Knijff, E.M.; Ruwhof, C.; de Wit, H.J.; Kupka, R.W.; Vonk, R.; Akkerhuis, G.W.; Nolen, W.A.; Drexhage, H.A. Monocyte-derived dendritic cells in bipolar disorder. Biol. Psychiatry 2006, 59, 317–326. [Google Scholar] [CrossRef] [PubMed]
  68. Ebstein, F.; Lange, N.; Urban, S.; Seifert, U.; Kruger, E.; Kloetzel, P.M. Maturation of human dendritic cells is accompanied by functional remodelling of the ubiquitin-proteasome system. Int. J. Biochem. Cell Biol. 2009, 41, 1205–1215. [Google Scholar] [CrossRef] [PubMed]
  69. Padmos, R.C.; Hillegers, M.H.; Knijff, E.M.; Vonk, R.; Bouvy, A.; Staal, F.J.; de Ridder, D.; Kupka, R.W.; Nolen, W.A.; Drexhage, H.A. A discriminating messenger RNA signature for bipolar disorder formed by an aberrant expression of inflammatory genes in monocytes. Arch. Gen. Psychiatry 2008, 65, 395–407. [Google Scholar] [CrossRef] [PubMed]
  70. Badner, J.A.; Koller, D.; Foroud, T.; Edenberg, H.; Nurnberger, J.I., Jr.; Zandi, P.P.; Willour, V.L.; McMahon, F.J.; Potash, J.B.; Hamshere, M.; et al. Genome-wide linkage analysis of 972 bipolar pedigrees using single-nucleotide polymorphisms. Mol. Psychiatry 2012, 17, 818–826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Tsuboi, D.; Kuroda, K.; Tanaka, M.; Namba, T.; Iizuka, Y.; Taya, S.; Shinoda, T.; Hikita, T.; Muraoka, S.; Iizuka, M.; et al. Disrupted-in-schizophrenia 1 regulates transport of ITPR1 mRNA for synaptic plasticity. Nat. Neurosci. 2015, 18, 698–707. [Google Scholar] [CrossRef] [PubMed]
  72. Mitchell, A.C.; Javidfar, B.; Pothula, V.; Ibi, D.; Shen, E.Y.; Peter, C.J.; Bicks, L.K.; Fehr, T.; Jiang, Y.; Brennand, K.J.; et al. MEF2C transcription factor is associated with the genetic and epigenetic risk architecture of schizophrenia and improves cognition in mice. Mol. Psychiatry 2017. [Google Scholar] [CrossRef] [PubMed]
  73. Hendriks, W.J.; Pulido, R. Protein tyrosine phosphatase variants in human hereditary disorders and disease susceptibilities. Biochim. Biophys. Acta 2013, 1832, 1673–1696. [Google Scholar] [CrossRef] [PubMed]
  74. Arey, R.N.; Enwright, J.F., 3rd; Spencer, S.M.; Falcon, E.; Ozburn, A.R.; Ghose, S.; Tamminga, C.; McClung, C.A. An important role for cholecystokinin, a CLOCK target gene, in the development and treatment of manic-like behaviors. Mol. Psychiatry 2014, 19, 342–350. [Google Scholar] [CrossRef] [PubMed]
  75. Spijker, A.T.; van Rossum, E.F.; Hoencamp, E.; DeRijk, R.H.; Haffmans, J.; Blom, M.; Manenschijn, L.; Koper, J.W.; Lamberts, S.W.; Zitman, F.G. Functional polymorphism of the glucocorticoid receptor gene associates with mania and hypomania in bipolar disorder. Bipolar Disord. 2009, 11, 95–101. [Google Scholar] [CrossRef] [PubMed]
  76. Spijker, S.; Van Zanten, J.S.; De Jong, S.; Penninx, B.W.; van Dyck, R.; Zitman, F.G.; Smit, J.H.; Ylstra, B.; Smit, A.B.; Hoogendijk, W.J. Stimulated gene expression profiles as a blood marker of major depressive disorder. Biol. Psychiatry 2010, 68, 179–186. [Google Scholar] [CrossRef] [PubMed]
  77. Azevedo, J.A.; Carter, B.S.; Meng, F.; Turner, D.L.; Dai, M.; Schatzberg, A.F.; Barchas, J.D.; Jones, E.G.; Bunney, W.E.; Myers, R.M.; et al. The microRNA network is altered in anterior cingulate cortex of patients with unipolar and bipolar depression. J. Psychiatr. Res. 2016, 82, 58–67. [Google Scholar] [CrossRef] [PubMed]
  78. Muhleisen, T.W.; Leber, M.; Schulze, T.G.; Strohmaier, J.; Degenhardt, F.; Treutlein, J.; Mattheisen, M.; Forstner, A.J.; Schumacher, J.; Breuer, R.; et al. Genome-wide association study reveals two new risk loci for bipolar disorder. Nat. Commun. 2014, 5, 3339. [Google Scholar] [CrossRef] [PubMed]
  79. Perlis, R.H.; Huang, J.; Purcell, S.; Fava, M.; Rush, A.J.; Sullivan, P.F.; Hamilton, S.P.; McMahon, F.J.; Schulze, T.G.; Potash, J.B.; et al. Genome-wide association study of suicide attempts in mood disorder patients. Am. J. Psychiatry 2010, 167, 1499–1507. [Google Scholar] [CrossRef] [PubMed]
  80. Gouvea, E.S.; Ota, V.K.; Noto, C.; Santoro, M.L.; Spindola, L.M.; Moretti, P.N.; Carvalho, C.M.; Xavier, G.; Rios, A.C.; Sato, J.R.; et al. Gene expression alterations related to mania and psychosis in peripheral blood of patients with a first episode of psychosis. Transl. Psychiatry 2016, 6, e908. [Google Scholar] [CrossRef] [PubMed]
  81. Pulay, A.J.; Rethelyi, J.M. Multimarker analysis suggests the involvement of BDNF signaling and microRNA biosynthesis in suicidal behavior. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 2016, 171, 763–776. [Google Scholar] [CrossRef] [PubMed]
  82. Shelley, W.B.; Shelley, E.D. A dermatologic diary. Portrait of a practice. Cutis 1992, 50, 179–186. [Google Scholar] [PubMed]
  83. Detera-Wadleigh, S.D.; Badner, J.A.; Berrettini, W.H.; Yoshikawa, T.; Goldin, L.R.; Turner, G.; Rollins, D.Y.; Moses, T.; Sanders, A.R.; Karkera, J.D.; et al. A high-density genome scan detects evidence for a bipolar-disorder susceptibility locus on 13q32 and other potential loci on 1q32 and 18p11.2. Proc. Natl. Acad. Sci. USA 1999, 96, 5604–5609. [Google Scholar] [CrossRef] [PubMed]
  84. Saito, T.; Guan, F.; Papolos, D.F.; Lau, S.; Klein, M.; Fann, C.S.; Lachman, H.M. Mutation analysis of SYNJ1: A possible candidate gene for chromosome 21q22-linked bipolar disorder. Mol. Psychiatry 2001, 6, 387–395. [Google Scholar] [CrossRef] [PubMed]
  85. McQuillin, A.; Bass, N.J.; Kalsi, G.; Lawrence, J.; Puri, V.; Choudhury, K.; Detera-Wadleigh, S.D.; Curtis, D.; Gurling, H.M. Fine mapping of a susceptibility locus for bipolar and genetically related unipolar affective disorders, to a region containing the C21ORF29 and TRPM2 genes on chromosome 21q22.3. Mol. Psychiatry 2006, 11, 134–142. [Google Scholar] [CrossRef] [PubMed]
  86. Ruderfer, D.M.; Fanous, A.H.; Ripke, S.; McQuillin, A.; Amdur, R.L.; Schizophrenia Working Group of the Psychiatric Genomics Consortium; Bipolar Disorder Working Group of the Psychiatric Genomics Consortium; Cross-Disorder Working Group of the Psychiatric Genomics Consortium; Gejman, P.V.; O’Donovan, M.C.; et al. Polygenic dissection of diagnosis and clinical dimensions of bipolar disorder and schizophrenia. Mol. Psychiatry 2014, 19, 1017–1024. [Google Scholar] [PubMed]
  87. Psychiatric GWAS Consortium Bipolar Disorder Working Group. Large-scale genome-wide association analysis of bipolar disorder identifies a new susceptibility locus near ODZ4. Nat. Genet. 2011, 43, 977–983. [Google Scholar] [Green Version]
  88. Ruzicka, W.B.; Subburaju, S.; Benes, F.M. Circuit- and Diagnosis-Specific DNA Methylation Changes at gamma-Aminobutyric Acid-Related Genes in Postmortem Human Hippocampus in Schizophrenia and Bipolar Disorder. JAMA Psychiatry 2015, 72, 541–551. [Google Scholar] [CrossRef] [PubMed]
  89. Benes, F.M.; Lim, B.; Matzilevich, D.; Walsh, J.P.; Subburaju, S.; Minns, M. Regulation of the GABA cell phenotype in hippocampus of schizophrenics and bipolars. Proc. Natl. Acad. Sci. USA 2007, 104, 10164–10169. [Google Scholar] [CrossRef] [PubMed]
  90. Noor, A.; Lionel, A.C.; Cohen-Woods, S.; Moghimi, N.; Rucker, J.; Fennell, A.; Thiruvahindrapuram, B.; Kaufman, L.; Degagne, B.; Wei, J.; et al. Copy number variant study of bipolar disorder in Canadian and UK populations implicates synaptic genes. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 2014, 165B, 303–313. [Google Scholar] [CrossRef] [PubMed]
  91. MacLaren, E.J.; Charlesworth, P.; Coba, M.P.; Grant, S.G. Knockdown of mental disorder susceptibility genes disrupts neuronal network physiology in vitro. Mol. Cell. Neurosci. 2011, 47, 93–99. [Google Scholar] [CrossRef] [PubMed]
  92. Hannah, J.; Zhou, P. Distinct and overlapping functions of the cullin E3 ligase scaffolding proteins CUL4A and CUL4B. Gene 2015, 573, 33–45. [Google Scholar] [CrossRef] [PubMed]
  93. Howell, K.R.; Floyd, K.; Law, A.J. PKBgamma/AKT3 loss-of-function causes learning and memory deficits and deregulation of AKT/mTORC2 signaling: Relevance for schizophrenia. PLoS ONE 2017, 12, e0175993. [Google Scholar] [CrossRef] [PubMed]
  94. Hicks, C.; Asfour, R.; Pannuti, A.; Miele, L. An integrative genomics approach to biomarker discovery in breast cancer. Cancer Inform. 2011, 10, 185–204. [Google Scholar] [CrossRef] [PubMed]
  95. Emamalizadeh, B.; Jamshidi, J.; Movafagh, A.; Ohadi, M.; Khaniani, M.S.; Kazeminasab, S.; Biglarian, A.; Taghavi, S.; Motallebi, M.; Fazeli, A.; et al. RIT2 Polymorphisms: Is There a Differential Association? Mol. Neurobiol. 2017, 54, 2234–2240. [Google Scholar] [CrossRef] [PubMed]
  96. Cross-Disorder Group of the Psychiatric Genomics Consortium. Identification of risk loci with shared effects on five major psychiatric disorders: A genome-wide analysis. Lancet 2013, 381, 1371–1379. [Google Scholar]
  97. Vakalopoulos, C. The effect of deficient muscarinic signaling on commonly reported biochemical effects in schizophrenia and convergence with genetic susceptibility loci in explaining symptom dimensions of psychosis. Front. Pharmacol. 2014, 5, 277. [Google Scholar] [CrossRef] [PubMed]
  98. Lo Vasco, V.R.; Longo, L.; Polonia, P. Phosphoinositide-specific Phospholipase C beta1 gene deletion in bipolar disorder affected patient. J. Cell Commun. Signal. 2013, 7, 25–29. [Google Scholar] [CrossRef] [PubMed]
  99. Ban, H.J.; Kim, S.C.; Seo, J.; Kang, H.B.; Choi, J.K. Genetic and metabolic characterization of insomnia. PLoS ONE 2011, 6, e18455. [Google Scholar] [CrossRef] [PubMed]
  100. Kanazawa, T.; Ikeda, M.; Glatt, S.J.; Tsutsumi, A.; Kikuyama, H.; Kawamura, Y.; Nishida, N.; Miyagawa, T.; Hashimoto, R.; Takeda, M.; et al. Genome-wide association study of atypical psychosis. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 2013, 162B, 679–686. [Google Scholar] [CrossRef] [PubMed]
  101. Yuan, P.; Zhou, R.; Wang, Y.; Li, X.; Li, J.; Chen, G.; Guitart, X.; Manji, H.K. Altered levels of extracellular signal-regulated kinase signaling proteins in postmortem frontal cortex of individuals with mood disorders and schizophrenia. J. Affect. Disord. 2010, 124, 164–169. [Google Scholar] [CrossRef] [PubMed]
  102. Kim, S.K.; Lee, J.Y.; Park, H.J.; Kim, J.W.; Chung, J.H. Association study between polymorphisms of the PARD3 gene and schizophrenia. Exp. Ther. Med. 2012, 3, 881–885. [Google Scholar] [CrossRef] [PubMed]
  103. Su, L.; Ling, W.; Jiang, J.; Hu, J.; Fan, J.; Guo, X.; Huang, G.; Xie, X.; Long, J. Association of EPHB1 rs11918092 and EFNB2 rs9520087 with psychopathological symptoms of schizophrenia in Chinese Zhuang and Han populations. Asia-Pac. Psychiatry 2016, 8, 306–308. [Google Scholar] [CrossRef] [PubMed]
  104. Chen, H.; Wang, N.; Zhao, X.; Ross, C.A.; O’Shea, K.S.; McInnis, M.G. Gene expression alterations in bipolar disorder postmortem brains. Bipolar Disord. 2013, 15, 177–187. [Google Scholar] [CrossRef] [PubMed]
  105. Geddes, J.R.; Miklowitz, D.J. Treatment of bipolar disorder. Lancet 2013, 381, 1672–1682. [Google Scholar] [CrossRef]
  106. Doherty, J.L.; Owen, M.J. Genomic insights into the overlap between psychiatric disorders: Implications for research and clinical practice. Genome Med. 2014, 6, 29. [Google Scholar] [CrossRef] [PubMed]
  107. Lee, S.A.; Tsao, T.T.; Yang, K.C.; Lin, H.; Kuo, Y.L.; Hsu, C.H.; Lee, W.K.; Huang, K.C.; Kao, C.Y. Construction and analysis of the protein–protein interaction networks for schizophrenia, bipolar disorder, and major depression. BMC Bioinform. 2011, 12, S20. [Google Scholar] [CrossRef] [PubMed]
  108. Fritzius, T.; Burkard, G.; Haas, E.; Heinrich, J.; Schweneker, M.; Bosse, M.; Zimmermann, S.; Frey, A.D.; Caelers, A.; Bachmann, A.S.; et al. A WD-FYVE protein binds to the kinases Akt and PKCzeta/lambda. Biochem. J. 2006, 399, 9–20. [Google Scholar] [CrossRef] [PubMed]
  109. Karege, F.; Perroud, N.; Schurhoff, F.; Meary, A.; Marillier, G.; Burkhardt, S.; Ballmann, E.; Fernandez, R.; Jamain, S.; Leboyer, M.; et al. Association of AKT1 gene variants and protein expression in both schizophrenia and bipolar disorder. Genes Brain Behav. 2010, 9, 503–511. [Google Scholar] [PubMed]
  110. Clarke, G.M.; Anderson, C.A.; Pettersson, F.H.; Cardon, L.R.; Morris, A.P.; Zondervan, K.T. Basic statistical analysis in genetic case–control studies. Nat. Protoc. 2011, 6, 121–133. [Google Scholar] [CrossRef] [PubMed]
  111. Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3000 shared controls. Nature 2007, 447, 661–678. [Google Scholar]
  112. Wellcome Trust Case Control Consortium. Available online: https://www.wtccc.org.uk/info/access_to_data_samples.html (accessed on 15 December 2017).
  113. Brown, G.R.; Hem, V.; Katz, K.S.; Ovetsky, M.; Wallin, C.; Ermolaeva, O.; Tolstoy, I.; Tatusova, T.; Pruitt, K.D.; Maglott, D.R.; et al. Gene: A gene-centered information resource at NCBI. Nucleic Acids Res. 2015, 43, D36–D42. [Google Scholar] [CrossRef] [PubMed]
  114. Renteria, M.E.; Cortes, A.; Medland, S.E. Using PLINK for Genome-Wide Association Studies (GWAS) and data analysis. Methods Mol. Biol. 2013, 1019, 193–213. [Google Scholar] [PubMed]
  115. Pathan, M.; Keerthikumar, S.; Ang, C.S.; Gangoda, L.; Quek, C.Y.; Williamson, N.A.; Mouradov, D.; Sieber, O.M.; Simpson, R.J.; Salim, A.; et al. FunRich: An open access standalone functional enrichment and interaction network analysis tool. Proteomics 2015, 15, 2597–2601. [Google Scholar] [CrossRef] [PubMed]
  116. Chen, J.; Bardes, E.E.; Aronow, B.J.; Jegga, A.G. ToppGene Suite for gene list enrichment analysis and candidate gene prioritization. Nucleic Acids Res. 2009, 37, W305–W311. [Google Scholar] [CrossRef] [PubMed]
  117. Szklarczyk, D.; Morris, J.H.; Cook, H.; Kuhn, M.; Wyder, S.; Simonovic, M.; Santos, A.; Doncheva, N.T.; Roth, A.; Bork, P.; et al. The STRING database in 2017: Quality-controlled protein–protein association networks, made broadly accessible. Nucleic Acids Res. 2017, 45, D362–D368. [Google Scholar] [CrossRef] [PubMed]
  118. Bader, G.D.; Hogue, C.W. An automated method for finding molecular complexes in large protein interaction networks. BMC Bioinform. 2003, 4, 2. [Google Scholar] [CrossRef] [Green Version]
  119. Kohl, M.; Wiese, S.; Warscheid, B. Cytoscape: Software for visualization and analysis of biological networks. Methods Mol. Biol. 2011, 696, 291–303. [Google Scholar] [PubMed]
  120. Barabasi, A.L.; Oltvai, Z.N. Network biology: Understanding the cell’s functional organization. Nat. Rev. Genet. 2004, 5, 101–113. [Google Scholar] [CrossRef] [PubMed]
  121. Schweiger, R.; Linial, M.; Linial, N. Generative probabilistic models for protein–protein interaction networks—The biclique perspective. Bioinformatics 2011, 27, i142–i148. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Results of the genome wide association study (GWAS). The horizontal axis represents 22 chromosomes and the vertical axis represents the negative logarithm with base 10 of GWAS p-value for each SNP. Red line: canonical 5 × 10−8 cutoff. Blue line: 0.01 cutoff used in this study.
Figure 1. Results of the genome wide association study (GWAS). The horizontal axis represents 22 chromosomes and the vertical axis represents the negative logarithm with base 10 of GWAS p-value for each SNP. Red line: canonical 5 × 10−8 cutoff. Blue line: 0.01 cutoff used in this study.
Ijms 18 02763 g001
Figure 2. Overlapped genes associated with four mental illnesses.
Figure 2. Overlapped genes associated with four mental illnesses.
Ijms 18 02763 g002
Figure 3. BD risk gene interaction network. Only the nodes with a degree ≥4 are shown. Green balls are BD risk genes identified in the GWAS with p < 0.01.
Figure 3. BD risk gene interaction network. Only the nodes with a degree ≥4 are shown. Green balls are BD risk genes identified in the GWAS with p < 0.01.
Ijms 18 02763 g003
Figure 4. The topology properties of the network. (a) The distribution of number of nodes with different degrees. (b) Frequency distribution of shortest paths. (c) The relationship between topological coefficients and the number of node neighbors. (d) The relationship between betweenness and the number of node neighbors.
Figure 4. The topology properties of the network. (a) The distribution of number of nodes with different degrees. (b) Frequency distribution of shortest paths. (c) The relationship between topological coefficients and the number of node neighbors. (d) The relationship between betweenness and the number of node neighbors.
Ijms 18 02763 g004
Figure 5. Gene clusters identified with Cytoscape. Yellow nodes are core hub genes. No core hub gene is found in Cluster 4 (d). (a) Cluster 1; (b) Cluster 2; (c) Cluster 3; (d) Cluster 4; (e) Cluster 5.
Figure 5. Gene clusters identified with Cytoscape. Yellow nodes are core hub genes. No core hub gene is found in Cluster 4 (d). (a) Cluster 1; (b) Cluster 2; (c) Cluster 3; (d) Cluster 4; (e) Cluster 5.
Ijms 18 02763 g005
Table 1. Molecular functions (GO).
Table 1. Molecular functions (GO).
NameFDRGene Count
transferase activity, transferring phosphorus-containing groups5.67 × 10−5118
kinase activity5.67 × 10−5103
phosphotransferase activity, alcohol group as acceptor5.67 × 10−596
protein serine/threonine kinase activity1.03 × 10463
protein kinase activity1.90 × 10481
GTPase regulator activity2.61 × 10447
signal transducer activity, downstream of receptor2.61 × 10433
GTPase activator activity4.57 × 10443
adenyl ribonucleotide binding7.16 × 104154
Table 2. Cellular components (GO).
Table 2. Cellular components (GO).
NameFDRGene Count
synapse5.23 × 10−14131
postsynapse1.68 × 10−1383
synapse part7.30 × 10−13110
synaptic membrane1.89 × 10−1262
cell junction9.04 × 10−10149
postsynaptic embrane1.91 × 10−947
neuron part2.94 × 10−9178
excitatory synapse7.86 × 10−949
plasma membrane region8.87 × 10−9130
neuron projection8.89 × 10−9144
Table 3. Biological processes (GO).
Table 3. Biological processes (GO).
NameFDRGene Count
neurogenesis1.28 × 10−7186
cell morphogenesis1.28 × 107159
generation of neurons1.28 × 107176
regulation of nervous system development1.28 × 107116
neuron differentiation3.55 × 107162
neuron development3.55 × 107136
cell projection morphogenesis3.61 × 107116
cellular component morphogenesis4.18 × 107164
cell projection organization4.63 × 107163
neuron projection morphogenesis6.25 × 10788
Table 4. The gene nodes with a network degree >17.
Table 4. The gene nodes with a network degree >17.
Hub GeneDegreeHub GeneDegreeHub GeneDegree
CDK162PNPLA6 [16]27FARS220
PTEN [17,18]61SYNJ227FBXO2220
BCL2 [19]60UBE2R2 [20]27FLT320
POLR2A [21]55CACNA1D [22,23,24]26GATA4 [25] *20
SMARCA2 [26,27]55CDK6 [28]26ITSN2 [29]20
GSK3B [30,31,32,33]54CHRM2 [34]26KIF18A20
ABL1 [35,36,37]53MTHFD1L [38] **26LONRF120
PRKCA [39,40]50GRIA1 [41]25NCOA320
bFGF [42] **48POLR2H25PCNT [43]20
RB1 [44] *45TJP1 [45] *25PJA220
KIT [11]40MAPRE1 [46] *24SYT1 [47]20
RAD51 *38RUNX1 [48]24TRIM3920
SIRT1 [49,50,51]38UBE2D424WDFY2 [52] **20
UBE2D1 [53,54]37EHHADH23AK419
DLG1 [55,56]36IQCB123ASB1519
CDC27 [57]35PPM1B [58]23ATF2 [29]19
NEDD4L [59]35PPP4C [60] *23BUB1B19
PRKG1 [61] *35RAD5023DHX1519
RAP1A34SH3GL223DNM3 [62] *19
CDH2 [63] *33DCTN122ETV619
GNB5 [64] *33ERBB4 [65,66]22FBXL13 [67,68] **19
MAPK6 [69]33FBXO3222HECW219
GNG7 [70]32ITPR1 [71] *22MEF2C [72] *19
PTPN11 [73] *32MLL [74]22NR3C1 [75]19
ZBTB16 [76]32NCOR2 [77]22BDE4D19
ADCY2 [78]31PRKCE [79]22RNF19B19
DICER1 [80,81]31RAD51B22RNF21719
SYNJ1 [82,83,84,85]31ACTN421RXFP219
CACNA1C [9,52,86,87]30CCND2 [88,89]21RYR119
CTTN30CDH521THBS219
DLG2 [90,91]30CUL4A [92] *21AKT3 [93] *18
MAP3K1 [94] *30EFCAB1321BARD118
RIT2 [95]30LMO721CTNNA2 [11,96]18
ANAPC5 [28]28MITF21HDAC718
PLCB1 [97,98,99]28TRIM9 [100]21ITGAV18
RAF1 [101] *28CCND320PARD3 [102] *18
PARK2 [61] *27EPHB1 [103] *20PCSK218
PLCG2 [13,39]27
* associated with other mental illness ** core hub genes.
Table 5. Significant risk gene modules.
Table 5. Significant risk gene modules.
ClusterScoreNodesEdgesNode IDs
12020190ASB15, HECW2, UBE2D1, NEDD4L, ANAPC5, PJA2
TRIM39, UBE2R2, UBE2D4, CDC27, TRIM9, ZBTB16
LONRF1, PARK2, FBXL13 *, FBXO22, RNF19B, LMO7
RNF217, FBXO32
26.12161SYNJ1, KIT, PIK3C2G, PTPN11, PIK3C2B, SYNJ2
RUNX1, ITSN2, PLCB1, CDH2, DNM3, SYT1, CTTN WDFY2 *, CHRM2, CCND2, MITF, PLCG2, CDK6,
ETV6, SH3GL2
35.51333MLL, bFGF(FGF2) *, BUB1B, BARD1, RB1, DICER1, RAD50, RAD51, BCL2, CDH5, SMARCA2, ABL1, CCND3
44.1821223AKT3, PTEN, ITPR1, PRKCE, GNB5, CDK1
ERBB4, GNG7, RAF1, GSK3B, PPM1B, MAP3K1
5333FARS2, RAD51B, MTHFD1L *
* core hub genes.

Share and Cite

MDPI and ACS Style

Xie, Z.; Yang, X.; Deng, X.; Ma, M.; Shu, K. A Genome-Wide Association Study and Complex Network Identify Four Core Hub Genes in Bipolar Disorder. Int. J. Mol. Sci. 2017, 18, 2763. https://doi.org/10.3390/ijms18122763

AMA Style

Xie Z, Yang X, Deng X, Ma M, Shu K. A Genome-Wide Association Study and Complex Network Identify Four Core Hub Genes in Bipolar Disorder. International Journal of Molecular Sciences. 2017; 18(12):2763. https://doi.org/10.3390/ijms18122763

Chicago/Turabian Style

Xie, Zengyan, Xianyan Yang, Xiaoya Deng, Mingyue Ma, and Kunxian Shu. 2017. "A Genome-Wide Association Study and Complex Network Identify Four Core Hub Genes in Bipolar Disorder" International Journal of Molecular Sciences 18, no. 12: 2763. https://doi.org/10.3390/ijms18122763

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop